Abstract
Extracellular vesicles (EVs) are indispensable mediators of intercellular communication, but they can also assume a nefarious role by ferrying pathological cargo that contributes to neurological, oncological, inflammatory, and infectious diseases. The canonical pathway for generating EVs involves the endosomal sorting complexes required for transport (ESCRT) machinery, but an alternative pathway is induced by the enrichment of lipid membrane ceramides generated by neutral sphingomyelinase 2 (nSMase2). Inhibition of nSMase2 has become an attractive therapeutic strategy for inhibiting EV biogenesis, and a growing number of small–molecule nSMase2 inhibitors have shown promising therapeutic activity in preclinical disease models. This review outlines the function of EVs, their potential role in disease, the discovery and efficacy of nSMase2 inhibitors, and the path to translate these findings into therapeutics.
Introduction: extracellular vesicles
EVs are small, membrane-enclosed vesicles ranging in size from 30 nm to several microns in diameter. Although the minimum size of an EV is defined by the physical constraints of membrane composition and curvature, the maximum size is limited by its biogenesis pathway [1]. Exosomes arise from the inward budding of the membrane of the multivesicular body (MVB) to form intraluminal vesicles (ILVs), which are released into the extracellular space as exosomes upon MVB–plasma membrane fusion [1] (Figure 1). Whereas exosomes are limited in size by the dimensions of the MVB, ectosomes, also called microvesicles or microparticles, bud outward from the plasma membrane. Ectosomes can be as small as exosomes, but can also reach multiple microns in diameter [2]. The isolation of specific EV subtypes is difficult, beause there are no reliable universal markers, and each has overlapping characteristics [3]. Each isolation technique has a bias toward different subsets of EVs and varying levels of non-EV contaminants, further complicating efforts to define EV populations (reviewed in [4]). Novel naming conventions instead focus on the general size or other physical characteristics of the EV, such as subdividing classes into small vesicles (<200 nm) and medium/large vesicles (>200 nm). In this review, we follow the guidelines established in [3] and use the general term ‘EV’.
Figure 1.
Schematic of extracellular vesicle (EV) biogenesis, cargo content, and physiological roles. EV biogenesis begins with the formation of interluminal vesicles (ILVs) to generate multivesicular bodies (MVBs) via either an endosomal sorting complexes required for transport (ESCRT)-dependent or -independent pathway. (a) ESCRT-dependent biogenesis requires the involvement of the ESCRT machinery to assist in the inward budding and pinching off of the ILVs. (b) ESCRT-independent biogenesis relies on increased ceramide concentrations generated by neutral Sphingomyelinase 2 (nSMase2), causing the membrane to bud inward to form ILVs. (c) MVBs contain many ILVs that can be targeted towards the plasma membrane for extracellular release. (d) The MVB fuses with the plasma membrane and releases the ILVs, which upon extracellular release become EVs. (e) EVs can carry a variety of cargo, both cytosolic and membrane associated, including lipids, proteins, and nucleic acids. (f) The biological consequences of EV release can be physiological, such as the elimination of cellular waste and cell-cell communication, or pathological, where disease-associated cargos contribute to cancer, inflammation, neurodegenerative disease, and infectious disease progression. Abbreviation: SM, sphingomyelin. Figure created with BioRender (BioRender.com).
The beginnings of modern EV research are often traced to important studies during the 1980s, when the groups of Trams, Johnstone, and Stahl discovered that cells use EVs to shed certain proteins [5-7]. However, hints of the diverse functionality of EVs in intercellular communication had been present in the literature for decades prior [8], ranging from involvement of platelet releasates in coagulation [9] to the membranous mating-type ‘gamones’ of Chlamydomonas [10] to bone EV-mediated calcification [11]. However, establishment of a strong field of study would wait until evidence emerged during the early 2000s that EVs could function similar to viruses in transferring genetic information between cells [12]. We now understand that EVs have broad and crucial roles in maintaining normal cell health and function through long- and short-range intercellular communication.
Extracellular vesicle biogenesis
EVs contain a variety of cargo, including proteins, nucleic acids, and bioactive lipids, which are packaged from the original cell and travel through the extracellular space, or broadly through bodily fluids, before being incorporated into recipient cells. In part because of the heterogeneity of the cargo, the mechanisms responsible for sorting specific cargo into EVs and how they are then targeted to appropriate distal destinations are not fully understood, and are an active area of research.
Diverse EV biogenesis mechanisms are found at different subcellular sites. There are two major pathways identified for EV biogenesis originating from the MVB. The first to be discovered was the ESCRT pathway, which comprises four distinct subcomplexes of ESCRT-0, -I, -II, and –III, together with the ATPase vacuolar protein sorting-associated protein 4 (VPS4). Each of these has distinct and sequential roles, including initiating ILV budding (-0), binding cargo (-I, and -II), vesicle maturation and constriction (-III), and membrane scission of the ILV (VPS4) [13]. Several subunits and accessory proteins are used as EV markers, such as ALG-2 interacting protein X (Alix), an ESCRT-III accessory protein that stabilizes the Snf7 subunit, and Tumor susceptibility gene 101 (Tsg101), an ESCRT-I subunit thought to bind ubiquitinated cargo[13]. In 2008, it was discovered that EVs can also be produced in the absence of ESCRT proteins, which led to the identification of a secondary, ceramide-dependent pathway [14]. A major source of ceramide is the hydrolysis of sphingomyelin (SM) into ceramide and phosphorylcholine by members of the sphingomyelinase (SMase) enzyme family (Figure 1a). Important biophysical properties of ceramide are its marked intrinsic negative curvature because of a small polar head group and its tendency to segregate into ceramide-enriched microdomains that facilitate the transition of the membrane from a more linear lamellar phase to a more densely packed and rounded hexagonal phase [15]. A focal enrichment of ceramide at the membrane facilitates the formation of ILVs [14]. More recently, evidence for a novel third pathway of ILV budding, independent from both the ESCRT and ceramide pathways, involving ADP ribosylation factor 6 (ARF6) and phospholipase D2 (PLD2), has also been reported [16]. EVs that originate from the plasma membrane come about via rearrangement of cytoskeletal components and recruitment of proteins, which aid in the outward budding and scission of the vesicle into the extracellular space [2].
The sphingomyelinase family
Sphingomyelinases are a diverse family of enzymes responsible for catalyzing the hydrolysis of SM to ceramide and phosphorylcholine. The primary classification of these enzymes is based on their optimal enzymatic pH, cation requirement, and localization, and include lysosomal acidic sphingomyelinase (L-aSMase), secreted zinc-dependent acidic sphingomyelinase (S-aSMase), alkaline sphingomyelinase (alkSMase), and magnesium-dependent and independent neutral sphingomyelinase (nSMase).
aSMase is encoded by the sphingomyelinase phosphodiesterase 1 (SMPD1) gene and was first described in 1963 by Shimon Gatt [17]. Functioning at an optimal pH of 5, aSMase is widely expressed in mammalian tissues and is either localized in the lysosome or secreted. There is an inherited mutant form of aSMase that causes toxic accumulation of SM, leading to a lysosomal storage disorder called Niemann–Pick disease.
alkSMase was first described in 1969 by Åke Nilsson [18]. The enzyme was initially isolated from human duodenal contents and pig intestinal homogenates, where it had the highest level of activity at an alkaline pH of 9. Despite its SMase activity, its amino acid sequence is dissimilar from that of other SMases and more closely matches the nucleotide pyrophosphatase/phosphodiesterase (NPP) family, hence its gene name is ENPP7 [19]. Its subcellular localization is mainly to the plasma membrane and Golgi apparatus, and its tissue expression is enriched in intestinal mucosa. AlkSMase has not been as extensively studied as the other SMases, but a decrease in its expression has been linked to gastrointestinal (GI) cancers and inflammatory bowel disease [19].
nSMase was first described in 1967 by Schneider and Kennedy as a Mg2+-dependent SMase optimally active at pH 7.4 isolated from spleens of patients with Niemann-Pick disease [20]. Researchers then identified additional Mg2+-dependent SMases from human and rat brain tissue [21]. To date, four cation-dependent isoforms of nSMases have been discovered: nSMase 1, 2, 3, and mitochondrial-associated nSMase (MA-nSMase), which are encoded by SMPD2–5, respectively [22,23]. Although a Mg2+-independent cytosolic form of nSMase was described, little is known about it other than it is activated by vitamin D3 in vitro [24]. Whereas nSMase2 has been extensively studied, less is known about nSMase 1, 3 and MA-nSMase. nSMase1 is expressed ubiquitously, with protein localization in the Golgi apparatus and endoplasmic reticulum (ER). Its physiological role remains unknown, because neither nSMase1 knockout (KO) nor overexpression significantly impacts lipid storage or metabolism [23]. nSMase3 does not share sequence homology with nSMase 1 or 2, and is highly expressed in skeletal and cardiac muscle [23]. Similar to nSMase1, its function is poorly elucidated. MA-nSMase was initially identified in zebrafish, but has since been observed in murine brain, testis, pancreas, and epididymis [22]. It is not currently known whether MA-nSMase is expressed in human tissues. In vitro overexpression results in increased SMase activity and ceramide production, indicating functional enzymatic activity, but little else is known at this time [22].
By contrast, nSMase2 has been widely studied. It is highly expressed in the plasma membrane and Golgi apparatus of many mammalian cell types, with highest expression in the brain. nSMase2 has been implicated in the cellular stress response, ceramide-mediated apoptosis, inflammation, and EV biogenesis [14,23]. Unlike nSMase1-KO mice, nSMase2-KO mice exhibit substantial reductions in SMase activity and have a dwarfism phenotype, with skeletal defects and growth hormone deficits [25]. The role of nSMase2 in bone structure is further supported by fro/fro mice, which have an autosomal recessive mutation in SMPD3, causing significant attenuation of nSMase activity and characteristics similar to those taht arise in autosomal recessive forms of osteogenesis imperfecta [26]. Based on evidence demonstrating a significant reduction in total SMase activity in both the KO and mutant nSMase2 animals, nSMase2 appears to the predominant source of nSMase in the body and, consequently, has the potential to serve as an effective therapeutic target in several disease areas where ceramide-mediated release of EVs has a key pathogenic role. Here, we provide an overview of the recent advances made in the field, in particular with respect to the therapeutic utility of nSMase2 inhibition.
Evolution of neutral sphingomyelinase 2 inhibitors
Background
Since the discovery of SMase activity in various mammalian tissues with neutral optimal pH, several structurally diverse molecules have been reported as inhibitors of this enzymatic activity. Most of the early work on nSMase inhibitors was conducted using mammalian tissues as sources of the enzyme. Given the lack of knowledge regarding the existence of nSMase isoforms at the time, these inhibitors were reported as nSMase inhibitors. However, in this review, these compounds are referred to as nSMase2 inhibitors given that the nSMase activity of these tissues is predominantly derived from this isoform. Several review articles have been written at different times to capture the progress made in the field of nSMase2 inhibitors [27-30]. Hence, this section focuses on the representative nSMase2 inhibitors (Table 1), including the latest generation of nSMase2 inhibitors with greater translational potential.
Table 1.
Representative nSMase2 inhibitors
| Compound | IC50 (mM) | Enzyme source | Mode of inhibition | Origin of discovery | Refs |
|---|---|---|---|---|---|
|
1 | Rat brain | Mixed | Microbial screening | [32] |
|
2.8 | Rat brain | Unknown | Sphingomyelin analog | [33] |
|
1.7 (Ki value) |
Bovine brain | Competitive | Ceramide analog | [34] |
|
1 | Rat brain | Noncompetitive | High-throughput screening | [35] |
|
5 | Human recombinant | Noncompetitive | High-throughput screening | [36] |
|
0.03 | Human recombinant | Noncompetitive | High-throughput screening | [37] |
|
0.3 | Human recombinant | Noncompetitive | High-throughput screening followed by medicinal chemistry efforts | [38] |
|
0.5 | Human recombinant | Noncompetitive | High-throughput screening followed by medicinal chemistry efforts | [39] |
Microbial products
Early efforts on the search of nSMase2 inhibitors concentrated on screening of extracts containing microbial products. Although most compounds identified through this approach were reported to be relatively weak inhibitors (IC50 >10 μM), scyphostatin 1 (Table 1) isolated from Trichopeziza mollissima displayed potent nSMase2 inhibition with an IC50 value of 1.0 μM using rat brain microsomes as an enzyme source [31,32]. In the subsequent biological evaluation, scyphostatin showed ~50-fold selectivity over aSMase and inhibited lipopolysaccharide (LPS)-induced production of prostaglandin E2 and IL-1β in human monocytes [31]. Despite the potent inhibitory activity and promising biological profile, the utility of scyphostatin was limited because of its poor aqueous solubility and chemical instability [32]. Efforts to develop synthetic derivatives of scyphostatin failed to identify nSMase2 inhibitors with comparable potency.
Sphingomyelin and ceramide derivatives
Another approach undertaken to identify nSMase2 inhibitors was to explore synthetic derivatives of sphingomyelin and ceramide, the substrate and product of nSMase2-catalyzed reactions. For instance, compound 2 (Table 1) is a sphingomyelin analog with a carbamate moiety as a replacement for the phosphodiester group. It inhibited nSMase2 from rat brain microsomes with an IC50 value of 2.8 μM [33]. Despite its sphingomyelin-like structure, no significant inhibition of aSMase and Bacillus cereus SMase was achieved by compound 2 at concentrations up to 100 μM [33]. KY3535 (compound 3, Table 1) is a ceramide analog where the fatty acid amide group is replaced by a thiourea group [34]. It was found to inhibit partially purified bovine brain nSMase2 in a competitive manner with a Ki value of 1.7 μM. However, its selectivity over aSMase was not reported.
Pharmacological effects of compound 2 and KY3535 were not investigated in vivo. One possible reason is the lack of desirable absorption, distribution, metabolism, and excretion (ADME) properties because of the presence of a long carbon chain contributing to the high lipophilicity of these compounds.
nSMase2 inhibitors identified from screening
Screening of compound libraries was another approach explored by many groups in an attempt to identify nonlipid-like nSMase2 inhibitors. GW4869 (compound 4, Table 1) was identified by a high-throughput assay using a partially purified rat brain nSMase as a source of enzyme and radiolabeled sphingomyelin as a substrate [35]. GW4869 was found to be a noncompetitive inhibitor, with an IC50 value of 1 μM and no inhibitory activity against aSMase. Since it was first reported, GW4869 has served as a tool compound in several preclinical studies as described later. However, GW4869 was not further developed as a clinical candidate or used as a molecular template for further structural optimization because of its highly lipophilic core structure unsuitable to serve as a tractable lead.
The first nSMase2 screening assay with human recombinant enzyme led to the discovery of cambinol (compound 5, Table 1) as a potent nSMase2 inhibitor [36]. It exhibited a noncompetitive mode of inhibition with an IC50 value of 7 μM. Cambinol was found to be equally potent (IC50 = 6 μM) against the rat nSMase2, enabling more rigorous cross-species translational studies.
The screening method that identified cambinol was fluorescence based and more practical than radiosubstrate-based methods that had been predominantly used in nSMase2 assays previously. Indeed, the new screening assay method was further miniaturized into a 1536-well format, enabling a screening of >350 000 compounds in collaboration with National Center for Advancing Translational Sciences (NCATS). These efforts led to the discovery of 2,6-dimethoxy-4-(5-phenyl-4-thiophen-2-yl-1H-imidazol-2-yl)-phenol (DPTIP, compound 6, Table 1) with an IC50 value of 30 nM [37], representing one of the most potent nSMase2 inhibitors identified to date. DPTIP was one of the first nSMase2 inhibitors evaluated for ADME properties. It showed metabolic stability in both mouse and human liver microsomes, but unfortunately exhibited poor oral bioavailability. However, following intraperitoneal injection (10 mg/kg) in mice, DPTIP achieved a brain-to-plasma ratio of 0.26, with brain levels above its IC50 value for 4 h post injection.
nSMase2 inhibitors derived from screening hits
Additional screening efforts using the same assay method followed by extensive medicinal chemistry efforts led to the discovery of phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)-carbamate (PDDC, compound 7, Table 1) [38], a noncompetitive nSMase2 inhibitor with an IC50 value of 300 nM. Although PDDC exhibited tenfold weaker potency over DPTIP, it showed excellent overall ADME properties. It was found to be stable in both mouse and human microsomes. Furthermore, it showed excellent oral bioavailability (F%=88) and brain penetration (a brain-to-plasma ratio of 0.6) in mice, providing the potential to serve as a valuable tool compound to investigate the therapeutic utility of nSMase2 inhibition in various preclinical models, including neurological disorders.
Another screening campaign of a ~70 compound library largely comprising fused indolines followed by structural optimization efforts, which led to the discovery of compound 8 (Table 1), a dual nSMase2 and acetylcholinesterase (AChE) inhibitor with IC50 values of 500 nM and 1.7 mM, respectively [39]. Following subcutaneous injection (20 mg/kg) in mice, the average brain level of compound 8 at the peak was 262 ng/g (~0.8 μM), exceeding its IC50 value for nSMase2.
The modular nature of compounds DPTIP, PDDC, and compound 8 can be exploited for further systematic structural optimization by fine-tuning various parts of the parent molecules. Indeed, preliminary efforts along this line have already been attempted for DPTIP and PDDC [40,41].
Conclusion
Judging from the latest progress, the tremendous efforts devoted to the discovery of nSMase2 inhibitors over the past decades have begun to bear fruit, with several structurally distinct inhibitors serving as tractable leads for further optimization. DPTIP, PDDC, and compound 8 are particularly promising because they have already shown utility as in vivo probe molecules, as described later. New nSMase2 inhibitors emerging from these leads should be well positioned to accelerate therapeutic development, given the growing evidence demonstrating the therapeutic utility of nSMase2 inhibition, as detailed in the following sections.
Utility of nSMase2 inhibition in neurodegenerative diseases
Neurodegenerative disease was one of the first areas where nSMase2 inhibition was explored as a therapeutic approach. Increased ceramide levels have been observed in patients with Alzheimer’s Disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS) [42]. New insights into the mechanisms underlying these diseases indicate that EV-mediated transmission of toxic proteins might be associated with disease propagation; thus, nSMase2 inhibition of EV biogenesis was proposed as a therapeutic strategy. Several studies using pharmacological inhibition or genetic knockdown have been instrumental in demonstrating the potential of nSMase2 inhibition for the treatment of neurodegenerative diseases, detailed herein (Table 2).
Table 2.
Utility of nSMase2 inhibition/KO in preclinical models of neurological disorders
| Disease | Model | Result of nSMase2 inhibition/KO | Refs |
|---|---|---|---|
| AD | 5XFAD mice | Decreased serum EV levels, brain ceramide levels, glial activation, tau phosphorylation, and Aβ plaque load, improved memory | [43] |
| fro;5XFAD mice | Reduction in EV release in brain and blood, decreased ceramide levels and Aβ plaque load | [44] | |
| 5XFAD mice | Improved body weight and memory | [41] | |
| AAV-tau or PS19 mice | Reduced tau spread from entorhinal cortex to hippocampus in AAV-tau mice; reduced hippocampal tau in PS19 mice | [45] | |
| Synapatosomes from human AD brain | Reduced EV release and tau spread | [46] | |
| HEK293T tau RD P301S cells; P301S + ICV IL-1β in mice | Reduced tau spread in vitro; decreased brain-derived EV levels and lowered tau levels in brain-derived EVs in vivo | [39] | |
| ALS/FTD | Primary rat motor neuron cells | Prevented NGF-induced motor neuron death | [54] |
| MS | Cuprizone-fed mice | Inhibited elevated posterior corpus callosum nSMase2 activity and ceramide levels, improved myelination status | [61] |
| DMD | mdx:nSMase2 DKO mice; H2K cells | Reduced anxiety behavior and muscle inflammation, enhanced muscle function in DKO mice; enhanced survival in H2K cells with inhibitor | [60] |
| PD | SH-SY5Y neuroblastoma cells | Decreased secretion of γ-synuclein | [49] |
| Decreased transfer of α-synuclein between cells | [50] | ||
| Midbrain slice cultures | Decreased EV release and IFN-γ/LPS-induced dopaminergic neurodegeneration | [51] | |
| Prion disease | GT1-7 hypothalamic cells | Reduced EV biogenesis and PrP packaging | [57,62] |
| Mov neuroglial cells | Decreased release of PrP | [58] |
Alzheimer’s disease
Recent evidence implicated EV-dependent propagation of amyloid β (Aβ) and hyperphosphorylated tau within the AD brain. These data led to a proposed role for nSMase2 inhibition in ameliorating AD pathogenesis. The first in vivo assessment of nSMase2 inhibition in AD mice occurred in the Bieberich laboratory [43], where systemic GW4869 administration in 5XFAD mice markedly reduced brain ceramide levels, EV release, brain Aβ1-42, and amyloid plaques. This pharmacological inhibition replicated the findings of their earlier genetic study, in which nSMase2-deficient fro/fro mice crossed with 5XFAD AD mice (fro;5XFAD) had reduced amyloid plaques, decreased EVs, and an improvement in fear-conditioning behavior compared with 5XFAD wild-type mice [44]. Recently, these findings were reproduced using the nSMase inhibitor PDDC, demonstrating drug-induced improvements in body weight and performance in contextual and cued fear-conditioning assays in 5XFAD mice [41].
The effect of nSMase2 inhibition on EV-mediated tau propagation has also been investigated. The Ikezu lab was the first to demonstrate the utility of inhibiting EV release to slow tau propagation in an adeno-associated virus (AAV) mouse model of rapid tau propagation. They reported that tau spreads along connectivity pathways in the brain via an EV-dependent mechanism, which was attenuated by either genetic knockdown or pharmacological inhibition of nSMase2 [45]. Further studies utilizing synaptosomes isolated from human AD brain and naïve recipient cells demonstrated that the nSMase2 inhibitors cambinol and GW4869 reduced EV production and limited the spread of tau between donor and recipient cells [46]. The same team developed a carbamate furoindoline compound (compound 8 in Table 1) with dual nSMase2/AChE inhibitory activity, which significantly reduced the levels of tau cargo in brain-derived EVs isolated from P301S AD mice challenged with an intracerebroventricular (ICV) injection of IL-1β [39]. These data are translationally relevant because the presence of phosphorylated tau, as well as Aβ, in neuronally derived EVs isolated from plasma from patients with AD were found to be a strong predictor of AD disease progression [47].
Parkinson’s disease
Mounting evidence demonstrates that α-synuclein, a main component of the abnormal Lewy body aggregates in PD, propagates interneuronally in the brain via EVs [48]. GW4869 was shown to inhibit the secretion of γ-synuclein, an upstream stimulator of α-synuclein seeding [49]. More recently, cambinol and nSMase2 knockdown were demonstrated to inhibit EV release and block the transfer of human α-synuclein from donor SH-SY5Y neuronal cells to co-cultured naïve recipient cells [50]. These findings were supported by a separate study showing that GW4869-mediated inhibition of EV release from rat brain slice cultures resulted in reduced cell death following interferon (IFN)-γ/lipopolysaccharide (LPS) treatment [51]. Additionally, clinical data demonstrated that higher α-synuclein levels in plasma-derived neuronal EVs in patients with PD were associated with higher risk for motor symptom worsening. These data also raise the possibility that EV contents could serve as an accessible biomarker for PD disease progression [52].
Amyotrophic lateral sclerosis and frontotemporal dementia
EV-mediated transport of pathological proteins has also been implicated in ALS and frontotemporal dementia (FTD). Both transactive response (TAR) DNA-binding protein 43 (TDP-43) and superoxide dismutase 1 (SOD1), the main aggregate-forming proteins in patients with ALS/FTD, have been identified as EV cargo, where they have demonstrated the ability to seed misfolding in naïve recipient cells [53]. Primary rat motor neurons overexpressing mutant SOD1G93A exhibited reduced cell death in response to nerve growth factor (NGF) and nitric oxide (NO) when simultaneously treated with nSMase2 inhibitors [54]. In models of TDP-43 inclusions, the results were less clear. Whereas TDP-43 was present in EVs released from hTDP-43-containing Neuro2a cells as well as in brains from patients with ALS [55], GW4869 treatment in Neuro2a cells caused TDP-43 inclusion increases, not decreases. In addition, GW4869-treated TDP-43A315T mice exhibited worse motor deficits, increased muscle denervation, and enhanced cognitive deficits [55]. These studies highlight the need for further experimental clarification of the role of nSMase2 in ALS/FTD.
Prion disease
Prion diseases have also garnered increased interest in the therapeutic potential of nSMase2 inhibitors. In fact, the ‘prion-like’ theory of neurodegenerative diseases arose from the discovery that seed-capable prion proteins are packaged into EVs that are able to induce misfolding in recipient cells [53,56]. Despite the early understanding of the importance of EVs, there have only been a few studies conducted with nSMase2 inhibitors in prion disease models. The Hill laboratory showed that GW4869 treatment and small interfering (si)RNA knockdown of nSMase2 in GT1–7 neuronal cells infected with mouse-adapted human prion protein (PrP) reduced the amount of both total PrP and disease-associated PrP [57]. A separate study confirmed that nSMase inhibition was capable of reducing the amount of EV-associated PrP [58]. Although additional work is needed, these initial studies demonstrate a possible role for nSMase2 inhibition in prion diseases.
Other degenerative diseases
nSMase2 inhibition has also been implicated in other degenerative diseases, such as multiple sclerosis (MS) and Duchenne muscular dystrophy (DMD). Elevated ceramide levels have been observed in patients with MS, with a recent report highlighting the pathogenic role of nSMase2 in oligodendrocytes by showing that cambinol promotes remyelination in a mouse model of MS [59]. In DMD, knocking out nSMase2 in a mouse model reduced inflammation, enhanced muscle function, and alleviated some anxiety-like behaviors [60]. Interestingly, the authors also observed increased EVs in the media of H2K DMD myoblasts compared with normal mouse myoblasts and prolonged survival of H2K myoblasts with increasing concentrations of GW4869, suggesting that EV release is detrimental.
Conclusion
Although much of the work investigating nSMase2 inhibitors in neurological disorders has focused on AD, a growing number of neurological disorders are now being linked to abnormal nSMase2 function. These promising data highlight the importance of identifying potent, brain-penetrable nSMase2 inhibitors that could be translated into clinical studies.
Utility of nSMase2 inhibition in cancer
Given the role of ceramide in inducing apoptosis and growth arrest, as well as the location of nSMase2 on chromosome 16q22.1, a region commonly lost in prostate and breast cancer, nSMase2 was originally thought to have a tumor-suppressor role (reviewed in [63]). Recently, however, a growing body of evidence has implicated EVs in propagating tumorigenesis, metastasis, drug resistance, and immunotherapy [63]. This has led to renewed interest in nSMase2 inhibitors as potential cancer therapeutics (Table 3).
Table 3.
Utility of nSMase2 inhibition/KO in preclinical models of cancer
| Cancer | Model | Result of nSMase2 inhibition | Refs |
|---|---|---|---|
| Breast | Cal51 cells | Decreased tumor cell senescence induced by paclitaxel | [76] |
| 4T1 cells in mice | Inhibited tumor growth and augmented anti-PD-L1 therapeutic effect | [84] | |
| Cervical | HeLa cells | Enhanced antimigration effects of mifepristone, decreased cell viability, and enhanced apoptosis | [70] |
| HT29 and FHC cells | Attenuated CRC-derived EV-mediated migration of normal colon epithelial cells | [74] | |
| CRC | LoVo cells in mice | Suppressed tumor growth | [68] |
| HT29 and HCT116 cells | Inhibited hypoxic EV-mediated migration and invasion | [69] | |
| SW480 cells | Decreased oxaliplatin resistance | [79] | |
| Duodenal | AZ-Pa7 cells | Decreased accumulation of toxic polyadenylate-binding protein 1 in EVs | [65] |
| Gastric | M2 macrophages and MFC cells; MFC cells in mice | Inhibited M2 macrophage-mediated migration; decreased MFC metastasis | [72] |
| SCG7901/ADR cells | Decreased doxorubicin resistance | [80] | |
| Lung | HMEC-1 and A549 cells | Inhibited EV containing tumor conditioned media-mediated transformation of epithelial cells to cancer-like phenotype | [64] |
| A549 cells and cancer associated fibroblasts | Inhibited EV release by CAFs, prevented epithelial to mesenchymal transition | [71] | |
| HBMECs and NCI-H446 cells | Decreased metastasis markers in SCLC cells | [73] | |
| Multiple myeloma | 5TGM1 cells in mice | Blocked bortezomib resistance | [78] |
| Oral | HSC-3-R and SCC-9-R cells | Ameliorated cisplatin resistance | [77] |
| Pancreatic | PANC-1 cells | Inhibited proliferation, migration, and chemokine expression | [66] |
| CAF and AsPC1 cells; CAF and AsPC1 cells in NOD-SCID mice | Decreased survival of PDAC cells; slowed tumor growth | [75] | |
| Prostate | C4-2B-R, CWR-R1-R, and LNCaP-R cells | Attenuated enzalutamide resistance | [81] |
| TRAMPC2 cells in mice | Inhibited tumor growth via nSMase2-null tumors | [85] | |
| Skin | B16BL6 cells in vitro and in mice | Inhibited EV release and decreased proliferation; suppressed tumor growth | [67] |
Tumorigenesis and metastasis
Tumor-derived EVs facilitate tumor metastasis and transfer their content from parent tumor cells to naïve cells, leading to malignant transformations. EVs from tumor-conditioned media in vitro can transform normal endothelial cells into a proangiogenic phenotype, an effect that can be reduced with nSMase2 inhibition [64]. Tumor EVs often carry proteins and miRNAs that encourage tumor growth and metastasis, such as polyadenylate-binding protein 1 and miR-21-5; nSMase2 inhibition dramatically reduces their release and, thus, their protumor influence [65,66]. Tumor-derived EVs have also been shown to increase proliferation and limit apoptosis in both melanoma and colon cancer cells, an effect that was reduced following GW4869 treatment [67,68]. In scratch assays, migration of colorectal cancer (CRC) cells was attenuated by nSMase2 inhibition [69], while nSMase2 inhibition enhanced the inhibitory effects of metapristone on the migration of cervical cancer cells [70]. Media from cancer-associated fibroblasts (CAFs) were found capable of inducing an epithelial-to-mesenchymal transition (EMT), a hallmark of metastasis, in several human lung cancer cell lines; the nSMase inhibitor GW4869 blocked EV release from fibroblasts and prevented the isolated media from inducing EMT in recipient cells [71]. Not only are tumor-derived EVs affected by nSMase2 inhibition, but EVs derived from tumor-associated macrophages, which can drive metastasis, are also reduced, with GW4869 treatment leading to reduced metastasis [72]. Small cell lung cancer (SCLC) typically metastasizes to the brain and it was determined that EVs released by brain microvascular endothelial cells promoted the expression of S100 A16, a marker associated with brain metastases, in SCLC cells [73]. Inhibiting EV release from brain microvascular cells prevented increased S100 A16 expression and reduced survival in SCLC cells in response to H2O2 [73]. EVs isolated from Ca2+-dependent activator protein for secretion 1 (CAPS1)-overexpressing CRC cells enhanced migration of normal fetal colonic mucosal cells, but GW4869 significantly blocked this migration [74].
Drug resistance
In addition to influencing tumorigenesis and metastases, tumor-derived EVs have also been implicated in the induction of drug resistance [63]. Tumor-derived EVs are capable of transferring drug-resistance mechanisms to sensitive cells in a handful of cancer cell types, and nSMase2 inhibition can abrogate this process. For example, CAFs have been shown to release EVs that promote gemcitabine resistance in pancreatic ductal adenocarcinoma cells; GW4869 was shown to block the EV release and limit the resistance [75]. An adverse effect of paclitaxel and other chemotherapeutic agents is inducing senescence, allowing tumor cells to evade apoptosis and thereby creating a reservoir of cells that can provide an environment encouraging metastasis or can reactivate and metastasize themselves. Paclitaxel-induced senescent breast cancer cells have enhanced EV release, but nSMase2 inhibition blocks this enhanced released and reduces senescence-associated β-galactosidase (SA-β-Gal) expression and cell survival [76]. In another study, cisplatin-resistant oral squamous cell carcinoma cells were shown to release EVs containing miRNA-21, which induces cisplatin resistance in naïve recipient cells. When GW4869 was used to block the release of EVs from donor cells, EV-recipient cells exhibited reduced resistance [77]. Drug resistance to bortezomib in a murine model of multiple myeloma was also alleviated with GW4869 [78]. In human CRC cells, inhibition of miRNA-19b-containing EVs with nSMase2 inhibition led to decreased oxaliplatin resistance and enhanced cell death [79]. EVs secreted by gastric cancer cells and carrying miR-105-5p were found to elicit doxorubicin resistance, whereas treating the parent cells with GW4869 limited the transfer of resistance [80]. Several prostate cancer cells lines that are resistant to the androgen receptor inhibitor enzalutamide were found to have increased EV release, and inhibiting this release with GW4869 enhanced cell death [81]. Although most publications implicate enhanced EV release and nSMase2 in drug resistance, some report the opposite. For example, in human breast cancer cells, enhancing the release of EVs containing the breast cancer resistant protein (BCRP) was found to reduce doxorubicin resistance [82], highlighting the importance of evaluating the role of EVs and nSMase2 inhibition in drug resistance in individual cancers and against specific chemotherapies.
Immunotherapy
Another important role for tumor-derived EVs is modulating the tumor immune response. One of the mechanisms by which tumors evade the immune system is by upregulating Programmed death-ligand 1 (PD-L1), a cell surface immune checkpoint ligand that binds to PD-1 on effector T cells and dampens their response. PD-L1 has been observed on tumor-derived EVs from several different cancers and is able to interact with effector T cells and dampen their antitumor response [83]. Co-treatment of GW4869 with anti-PD-L1 antibody therapy improved the antitumor efficacy versus anti-PD-L1 antibody alone in a breast cancer model [84]. In a recent study by Poggio et al., the removal of EV PD-L1 via CRISPR/Cas9 inhibited tumor growth in the TRAMP-C2 murine model of prostate cancer [85]. Furthermore, injection of PD-L1-null or nSMase2-null TRAMP-C2 cells failed to generate tumors in mice, whereas injection of PD-L1-null TRAMP-C2 cells promoted T cell activation [85]. Remarkably, when WT TRAMP-C2 cells were injected in one flank of a mouse simultaneously with PD-L1-null or nSMase2-null TRAMP-C2 cells on the other flank, the wild-type tumors failed to grow, supporting the idea that immune cells activated on the PD-L1 or nSMase2-null side were able to migrate and attack the wild-type tumor cells on the opposite flank. These data suggest nSMase2 inhibition as an effective therapeutic combination with immunotherapy to enhance systemic antitumor immunity.
Conclusion
The role of nSMase2-dependent EVs in cancer is complex and varies between cancers, cargo content, and chemotherapies. Whereas some studies demonstrated a beneficial role nSMase2 in limiting cancer progression, the majority of literature points to nSMase2-mediated EV biogenesis as a mediator of tumorigenesis, metastasis, and drug resistance, with a role in diminishing antitumor immunity response, making it a logical therapeutic target in several cancers.
Utility of nSMase2 inhibition in inflammation
Over the past decade, the role of EVs in exacerbating inflammatory responses to injury and disease has gained attention, and the effectiveness of nSMase2-mediated inhibition of EV biogenesis and release on these conditions is just beginning to be explored (Table 4).
Table 4.
Utility of nSMase2 inhibition in preclinical models of inflammatory diseases
| Disease | Model | Result of nSMase2 inhibition | Refs |
|---|---|---|---|
| Airway inflammation | Intratracheal LPS | Improved lung elastance, decreased inflammation | [88] |
| Allergic airway inflammation | BEAS-2B cells; ovalbumin-sensitized mice | Decreased EV secretion in lung cells and lung inflammation | [86] |
| House dust mite-sensitized mice | Decreased airway-secreted EVs, Th2 cytokines, and eosinophils | [87] | |
| Atherosclerosis | Mesenteric small artery extracts | Decreased markers of vascular inflammation (VCAM) | [94] |
| DC-HUVEC co-culture | Decreased EV release and activation of HUVECs | [95] | |
| APOE−/− mice | Decreased atherosclerotic lesions, macrophage infiltration, and lipid deposition | [96] | |
| LDLR−/− mice | Reduced atherosclerosis, glucose, lipids, insulin resistance, and hepatic steatosis | [97] | |
| Cerebral ischemia | Focal cerebral ischemia | Inhibited EV release and decreased neuroinflammation | [90] |
| Transient global ischemia | Decreased hippocampal ceramide levels, neuronal damage, and inflammation | [91] | |
| Hepatic IR injury | Rat hepatic IR injury | Decreased liver COX activity and PGE2 levels | [89] |
| Myocardial infarction | Myocardial infarction | Reduced tachypnea, diaphragm dysfunction, and ceramide levels | [92] |
| Sepsis | Cecal ligation puncture | Decreased calpain activation and diaphragm weakness | [98] |
| Endotoxin or cecal ligation puncture | Inhibited EV release, decreased inflammation, and prolonged survival | [93] |
Inflammatory airway diseases
EVs are involved in the inflammatory response to airway diseases. For example, EVs isolated from bronchial epithelial cells have been shown to induce monocyte chemotaxis and proliferation [86]. In a mouse model of allergic airway inflammation, treatment with the nSMase2 inhibitor GW4869 led to fewer lung macrophages and improved airway hyper-responsiveness and bronchial pathology [86]. In house dust mite-sensitized mice, nSMase2 inhibition reduced the number of EVs in bronchoalveolar lavage fluid, resulting in fewer eosinophils in the lungs as well as declines in interleukin (IL)-4 and IL-13 [87]. In an LPS-induced mouse model of airway inflammation and injury, blockade of nSMase2 also reduced proinflammatory cytokines, IL-6 and IL-1β, and improved lung elastance, alveolar collapse, and immune cell infiltration, but whether these findings were the result of reduced EV release was not explored [88].
Ischemia-reperfusion injury and sepsis
Whereas aSMase has historically been the focus in ischemia-reperfusion (IR) injury, several publications report that inhibiting nSMase2 can also improve outcomes. In a liver IR model, GW4869 improved histopathological scores and decreased cyclooxygenase and prostaglandin E2 [89]. In preclinical cerebral ischemia models, blocking proinflammatory EV release from brain tissue with GW4869 resulted in fewer Iba1+ cells in the cortex and hippocampus and a shift in microglia from the proinflammatory state to anti-inflammatory state, as measured by a decrease in CD86 and increase in CD206 levels [90] and a reduction in inflammatory markers [91]. nSMase2 inhibition was also shown to reduce tachypnea and diaphragm dysfunction in a myocardial infarction model [92]. nSMase2-mediated EV release has also been implicated in the ‘cytokine storm’ phenomenon seen in sepsis. EVs isolated from LPS-stimulated macrophages were shown to contain enhanced levels of proinflammatory cytokines, which were capable of inducing further cytokine release from naïve macrophages [93]. In preclinical LPS-induced inflammation and cecal ligation puncture sepsis models, nSMase2 inhibition reduced myocardial inflammation and proinflammatory cytokine levels, improved cardiac function, and prolonged survival
Atherosclerosis
Chronic endothelial inflammation is implicated is atherosclerosis. In patients with hypertension, endothelin-1 is elevated and activates nSMase2, which increases vascular cell adhesion protein 1 (VCAM-1) and vascular inflammation, leading to small artery remodeling [94]. Inhibiting nSMase2 with GW4869 lowers VCAM-1 expression in rat mesenteric small arteries [94]. In addition to nSMase2 itself playing a part in vascular inflammation, EVs released from dendritic cells (DCs) contain tumor necrosis factor (TNF)-α and are capable of inducing activation of cultured human umbilical vein endothelial cells (HUVECs) via nuclear factor (NF)-κB signaling [95]. When DC EVs were inhibited by nSMase2 inhibition, the conditioned media could no longer activate endothelial cells, thereby significantly reducing levels of VCAM-1, intercellular cell adhesion protein 1 (ICAM-1), and E-selectin [95]. In addition to reducing pathogenic EVs, nSMase2 inhibition attenuated inflammatory responses and beneficially curtailed endothelial cell activation in vitro. This approach also decreased atherosclerotic lesion area in APOE−/− mice, an effect observed similarly following genetic depletion of nSMase2 or treatment with GW4869 [96]. Atherosclerotic plaque formation was also attenuated with nSMase2 inhibition in mice fed a high-fat diet before LPS treatment [97]. Interestingly, this same study also observed atherosclerotic plaque decreases with aSMase inhibition [97].
Conclusion
Taken together, there is mounting evidence that nSMase2 inhibition has a role in modulating multiple aspects of the inflammatory response and its inhibition might have therapeutic applications for myriad inflammatory diseases.
nSMase2 in infectious diseases
Inhibition of nSMase2 is being explored as a therapeutic target for bacterial and viral infections. Both nSMase2 and EVs in general have been implicated in multiple infectious diseases, including HIV, Zika virus, rabies virus, Dengue virus (DENV), and Shiga toxin (Table 5).
Table 5.
Utility of nSMase2 inhibition in preclinical infectious disease models
| Disease | Model | Result of nSMase2 inhibition | Refs |
|---|---|---|---|
| CMV | Primary human dermal fibroblasts | Decreased propagation of HCMV | [108] |
| DENV | C6/36 cells | Decreased EVs and viral load | [106] |
| Epsilon toxin | ACHN cells | Inhibited toxin-mediated ceramide formation and cytotoxicity | [110] |
| HCV | Huh7-Lunet-TLR3 cells | Decreased secretion of infectious virus | [103] |
| HEV | PLC/PRF/5 cells | Decreased secretion of infectious virus | [104] |
| HIV | DCs and CD4+ T cells | Blocked trans-infection from HIV-1-infected cells to healthy DCs and CD4+ T lymphocytes | [100] |
| Langat virus | ISE6 and HaCaT cells | Inhibited release of tick cell-derived EVs and decreased viral RNA transmission | [105] |
| NDV | DF-1 cells | Decreased EV release from infected cells and decreased extracellular levels of virus | [109] |
| Rabies virus | Vero cells | Decreased levels of viral RNA | [107] |
| Shiga toxin | THP-1 cells | Decreased toxin-mediated human renal cell death | [111] |
| Zika virus | Human fetal astrocytes | Decreased EV release and suppressed virus propagation | [101] |
| Primary murine cortical neurons | Reduced EV release and diminished viral RNA levels | [102] |
Viral infections
EVs are implicated in the propagation of HIV infection (reviewed in [99]). EVs released from HIV-infected cells carry HIV accessory proteins and co-receptors that make target cells more receptive to HIV infection. Additionally, the virion can physically associate with EVs, which can enable it to evade immune surveillance and increase infectivity. EVs from HIV-1-infected CD4+ T cells can also induce HIV-1 reactivation from dormant viral reservoirs in resting CD4+ T lymphocytes [100]. In cell culture experiments, blocking the release of EVs from infected CD4+ T cells with the nSMase2 inhibitors GW4869 and spiroepoxide reduced DC-mediated infection of healthy CD4+ T lymphocytes [100].
In addition to HIV, nSMase2 inhibitors have also shown therapeutic promise against the Zika virus. Zika infection in human fetal astrocytes increased the release of EVs and viral particles; some of the viral particles were packaged within the EVs. Inhibiting EV release via GW4869 led to diminished Zika virus propagation [101]. Similar findings were observed in murine neuronal cell cultures, where Zika virus led to enhanced EV release containing viral RNA. Either silencing nSMase2 using siRNA or pharmacologically inhibiting the enzyme with GW4869 reduced EV release and diminished viral RNA levels [102].
The efficacy of nSMase2 inhibitors has also been explored in Hepatitis C (HCV), Hepatitis E (HEV), Rabies, Langat virus (LGTV), DENV, cytomegalovirus (HCMV), and Newcastle disease virus (NDV). In HCV, double-stranded RNA was found in EVs released from infected hepatocytes; blocking EV release using GW4869 resulted in reduced viral replication [103]. Similarly, HEV particles were packaged into EVs and released from infected liver cells. Again, the EV release of HEV was decreased following nSMase2 inhibition [104]. A tick-borne flavivirus, LGTV, which is similar to tick-borne encephalitis virus, was found within EVs from both arthropod and mammalian cells. Inhibiting EV release lessened EV viral RNA loading and viral plaque formation [105]. Infectious RNA of another flavivirus, the mosquito-borne DENV, was also found in EVs released from DENV-infected mosquito-derived cell lines, and treatment with GW4869 partially blocked transmission to human cells [106]. Similarly, extracellular viral RNA, likely associated with EVs from rabies virus-infected cells, was also reduced following GW4869 treatment [107]. The human herpes virus, HCMV, was also found to utilize EVs for propagation, which was slowed by nSMase2 inhibition [108]. Interestingly, viral proteins packaged into EVs are not unique to mammalian infectious diseases because proteins of NDV, a poultry virus, were detected in EVs released from infected chicken fibroblasts, and GW4869 treatment lowered infectivity [109].
Bacterial infections
The effects of nSMase2 inhibition on bacterial infections are less studied than for viral infections, but a few publications have demonstrated utility. Epsilon-toxin produced by Clostridium perfringens, a lethal bacterial infection of undulates, was shown to enhance ceramide production in exposed kidney cells. Treatment of the exposed kidney cells with GW4869 reduced cell death [110]. The bacterial Shiga toxin, released by certain strains of Escherichia coli associated with GI, kidney, and central nervous system (CNS) pathology, was found to be packaged into EVs derived from exposed macrophages. These EVs induced cell death in naïve HK-2 renal epithelial cells. Renal epithelial cell death rates were ameliorated when EV release was blocked with nSMase2 inhibition [111].
Conclusion
There are strong indications that EVs are involved in the propagation and pathogenesis of viral infections, while preliminary evidence suggests their involvement in bacterial toxin-induced pathology. These areas represent exciting and possibly wide-reaching therapeutic applications for nSMase2 inhibitors.
Concluding remarks and future directions
This review summarizes the rapidly developing field of ceramide-dependent EVs as they relate to disease, with an emphasis on therapeutic opportunities afforded by nSMase2 inhibition. Although there is expert consensus that EVs contribute to the pathogenesis of a variety of diseases, caution must be exerted in unraveling the diverse biological networks that these EVs influence. First, EV isolation techniques are not standardized, and each technique demonstrates a bias toward certain subpopulations of EVs and varying degrees of non-EV contaminants [4]. As such, it is important to keep in mind the isolation technique used and any biases that might confound the results of the study when interpreting results. Second, several reports point to a beneficial role for ceramide-dependent EVs in diseases such as ALS [55], retinal degeneration [112], inflammatory bowel disease [113], and AD [114], which are in direct opposition to the mostly pathological role of these EVs reviewed earlier. It is probable that cell and cargo type, along with the stage of disease, dictates whether EVs are helpful or harmful, further complicating the application of nSMase2 inhibitors as therapeutic modalities. Adding to this, EVs isolated from umbilical cord mesenchymal stem cells have beneficial anti-inflammatory effects, highlighting both their role in maintaining homeostasis and their potential to be utilized as immunomodulatory therapeutics [115]. As EV separation and characterization techniques improve [116], we will be able to better assess their involvement in health and disease.
EVs also have a central role in neurodevelopmental processes [117] and nSMase2-KO mice have disrupted hypothalamic–pituitary–adrenal axes, leading to dwarfism, defects in bone formation, and overall hypoplasia [25]. These data warn that administration of nSMase2 inhibitors during childhood could be detrimental. In normal mice, one report showed that inhibition of nSMase2 worsened cognitive function [118], suggesting that disruption of EV function when there is no disease burden causes toxicity. Overall, there is much work to be done before nSMase2 inhibitors are available in the clinic, but existing research suggests that initial utility should be limited to mature adults diagnosed with diseases in which there is clear evidence that EVs contribute to pathological burden.
Although numerous nSMase2 inhibitors and genetic knockdown studies have demonstrated benefit in preclinical animal models of disease, the major impediment to clinical translation is the lack of a potent, drug-like inhibitor with good pharmacokinetic and safety profiles. Better compounds will also provide the ability to further understand the often complicated roles of EVs in disease. Given several exciting recent advances, such as the discovery of PDDC and DPTIP [37,38,40,41], there is optimism that a clinic-ready nSMase2 inhibitor is near, and we are buoyed with the knowledge that a growing number of research teams are engaged in exploring the therapeutic applications of this fascinating enzyme.
Research highlights:
Extracellular vesicles (EVs) are potent vehicles of intercellular communication
EVs can transport pathological cargo that contributes to disease
One pathway of EV biogenesis is dependent upon ceramides generated by nSMase2
Inhibition of nSMase2 shows promise in treating diseases that propagate via EVs
Potent and selective nSMase2 inhibitors have recently been discovered
Biography

Carolyn Tallon completed her BSc in physiology at McGill University before earning her PhD in pathobiology at Johns Hopkins University. She completed her doctoral thesis work in the lab of Mohamed Farah determining the efficacy of BACE1 inhibitors on enhancing peripheral nerve regeneration in injury and disease. She is currently a postdoctoral fellow in the lab of Barbara Slusher at Johns Hopkins University studying the efficacy of small-molecule inhibitors in neurodegenerative diseases, particularly neutral sphingomyelinase 2 inhibitors in Alzheimer’s disease.

Barbara Slusher is a professor of neurology, pharmacology, psychiatry, neuroscience, medicine and oncology at Johns Hopkins School of Medicine and the Director of Johns Hopkins Drug Discovery. Before joining Johns Hopkins, Dr Slusher spent 18 years in the pharmaceutical industry, and has extensive experience in drug discovery through early clinical development. In 2009, she joined Hopkins to lead a small-molecule drug discovery program with a veteran team of over 25 medicinal chemists, assay developers, pharmacologists/toxicologists, and pharmacokinetics/drug metabolism experts. The team is engaged in identifying novel drug targets and translating them into new drug therapies for clinical development.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of interests
C.T., B.S.S., R.R., T.T. and N.J.H. are listed as inventors in patent applications filed by Johns Hopkins Technology Ventures covering novel compositions of nSMase2 inhibitors and their utility.
References
- 1.Russell AE et al. (2019) Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: an ISEV position paper arising from the ISEV membranes and EVs workshop. Journal of Extracellular Vesicles 8, 1684862–1684862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tricarico C et al. (2017) Biology and biogenesis of shed microvesicles. Small GTPases 8, 220–232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Théry C et al. (2018) Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles 7, 1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Monguió-Tortajada M et al. (2019) Extracellular vesicle isolation methods: rising impact of size-exclusion chromatography. Cellular and Molecular Life Sciences 76, 2369–2382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pan B-T and Johnstone RM (1983) Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 33, 967–978 [DOI] [PubMed] [Google Scholar]
- 6.Trams EG et al. (1981) Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochimica et Biophysica Acta Biomembranes 645, 63–70 [DOI] [PubMed] [Google Scholar]
- 7.Harding C et al. (1984) Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol 35, 256–263 [PubMed] [Google Scholar]
- 8.Witwer KW and Théry C (2019) Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. Journal of Extracellular Vesicles 8, 1648167–1648167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wolf P (1967) The nature and significance of platelet products in human plasma. Br J Haematol 13, 269–288 [DOI] [PubMed] [Google Scholar]
- 10.Snell WJ (1976) Mating in Chlamydomonas: a system for the study of specific cell adhesion. I. Ultrastructural and electrophoretic analyses of flagellar surface components involved in adhesion. Journal of Cell Biology 68, 48–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Anderson HC (1969) Vesicles associated with calcification in the matrix of epiphyseal cartilage. The Journal of Cell Biology 41, 59–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lee Y et al. (2012) Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Human Molecular Genetics 21 (R1), R125–R134 [DOI] [PubMed] [Google Scholar]
- 13.Henne William M. et al. (2011) The ESCRT pathway. Developmental Cell 21, 77–91 [DOI] [PubMed] [Google Scholar]
- 14.Trajkovic K et al. (2008) Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319 (5867), 1244–1247 [DOI] [PubMed] [Google Scholar]
- 15.López-Montero I et al. (2007) Surface tension induced by sphingomyelin to ceramide conversion in lipid membranes. Biochimica et Biophysica Acta Biomembranes 1768, 553–561 [DOI] [PubMed] [Google Scholar]
- 16.Ghossoub R et al. (2014) Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat Commun 5, 3477. [DOI] [PubMed] [Google Scholar]
- 17.Gatt S (1963) Enzymic hydrolysis and synthesis of ceramides. Journal of Biological Chemistry 238, PC3131–PC3133 [PubMed] [Google Scholar]
- 18.Nilsson Å (1969) The presence of sphingomyelin- and ceramide-cleaving enzymes in the small intestinal tract. Biochimica et Biophysica Acta Lipids and Lipid Metabolism 176, 339–347 [DOI] [PubMed] [Google Scholar]
- 19.Duan R-D (2006) Alkaline sphingomyelinase: an old enzyme with novel implications. Biochimica et Biophysica Acta Molecular and Cell Biology of Lipids 1761, 281–291 [DOI] [PubMed] [Google Scholar]
- 20.Schneider PB and Kennedy EP (1967) Sphingomyelinase in normal human spleens and in spleens from subjects with Niemann-Pick disease. J Lipid Res 8, 202–209 [PubMed] [Google Scholar]
- 21.Gatt S (1976) Magnesium-dependent sphingomyelinase. Biochemical and Biophysical Research Communications 68, 235–241 [DOI] [PubMed] [Google Scholar]
- 22.Wu BX et al. (2010) Identification and characterization of murine mitochondria-associated neutral sphingomyelinase (MA-nSMase), the mammalian sphingomyelin phosphodiesterase 5. Journal of Biological Chemistry 285 (23), 17993–18002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wu BX et al. (2010) Mammalian neutral sphingomyelinases: regulation and roles in cell signaling responses. Neuromolecular Med 12, 320–330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Okazaki T et al. (1994) Characteristics and partial purification of a novel cytosolic, magnesium-independent, neutral sphingomyelinase activated in the early signal transduction of 1 alpha,25-dihydroxyvitamin D3-induced HL-60 cell differentiation. Journal of Biological Chemistry 269, 4070–077 [PubMed] [Google Scholar]
- 25.Stoffel W et al. (2005) Neutral sphingomyelinase 2 (smpd3) in the control of postnatal growth and development. Proc Natl Acad Sci U S A 102, 4554–559 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Aubin I et al. (2005) A deletion in the gene encoding sphingomyelin phosphodiesterase 3 (Smpd3) results in osteogenesis and dentinogenesis imperfecta in the mouse. Nature Genetics 37, 803–805 [DOI] [PubMed] [Google Scholar]
- 27.Delgado A et al. (2006) Inhibitors of sphingolipid metabolism enzymes. Biochim Biophys Acta 1758, 1957–1977 [DOI] [PubMed] [Google Scholar]
- 28.Skácel J et al. (2021) Small molecule inhibitors targeting biosynthesis of ceramide, the central hub of the sphingolipid network. J Med Chem 64, 279–297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Adada M et al. (2016) Inhibitors of the sphingomyelin cycle: sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids 197, 45–59 [DOI] [PubMed] [Google Scholar]
- 30.Bhabak KP and Arenz C (2013) Novel drugs targeting sphingolipid metabolism. Handb Exp Pharmacol 215, 187–196 [DOI] [PubMed] [Google Scholar]
- 31.Nara F et al. (1999) Biological activities of scyphostatin, a neutral sphingomyelinase inhibitor from a discomycete, Trichopeziza mollissima. J Antibiot (Tokyo) 52, 531–535 [DOI] [PubMed] [Google Scholar]
- 32.Nara F et al. (1999) Scyphostatin, a neutral sphingomyelinase inhibitor from a discomycete, Trichopeziza mollissima: taxonomy of the producing organism, fermentation, isolation, and physico-chemical properties. J Antibiot (Tokyo) 52, 525–530 [DOI] [PubMed] [Google Scholar]
- 33.Taguchi M et al. (2003) Sphingomyelin analogues as inhibitors of sphingomyelinase. Bioorganic & Medicinal Chemistry Letters 13, 1963–1966 [DOI] [PubMed] [Google Scholar]
- 34.Jung SM et al. (2003) Inhibition of a neutral form of sphingomyelinase by alkylthioureido-l,3-propandiols, KY353X series. J. Appl. Pharmacol 11, 169–173 [Google Scholar]
- 35.Luberto C et al. (2002) Inhibition of tumor necrosis factor-induced cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase. J Biol Chem 277 (43), 41128–41139 [DOI] [PubMed] [Google Scholar]
- 36.Figuera-Losada M et al. (2015) Cambinol, a novel inhibitor of neutral sphingomyelinase 2 shows neuroprotective properties. PLoS ONE 10, e0124481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rojas C et al. (2018) DPTIP, a newly identified potent brain penetrant neutral sphingomyelinase 2 inhibitor, regulates astrocyte–peripheral immune communication following brain inflammation. Scientific Reports 8, 17715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rojas C et al. (2019) A novel and potent brain penetrant inhibitor of extracellular vesicle release. Br J Pharmacol 176 (19), 3857–3870 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bilousova T et al. (2020) Dual neutral sphingomyelinase-2/acetylcholinesterase inhibitors for the treatment of Alzheimer’s disease. ACS Chemical Biology 15, 1671–1684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Stepanek O et al. (2019) Neutral sphingomyelinase 2 inhibitors based on the 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol scaffold. European Journal of Medicinal Chemistry 170, 276–289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sala M et al. (2020) Novel human neutral sphingomyelinase 2 inhibitors as potential therapeutics for Alzheimer disease. Journal of Medicinal Chemistry 63, 6028–6056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Alessenko AV and Albi E (2020) Exploring sphingolipid implications in neurodegeneration. Frontiers in Neurology 11, 437–437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dinkins MB et al. (2014) Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging 35, 1792–1800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dinkins MB et al. (2016) Neutral sphingomyelinase-2 deficiency ameliorates Alzheimer’s disease pathology and improves cognition in the 5XFAD mouse. J Neurosci 36 (33), 8653–8667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Asai H et al. (2015) Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 18, 1584–1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bilousova T et al. (2018) Suppression of tau propagation using an inhibitor that targets the DK-switch of nSMase2. Biochemical and Biophysical Research Communications 499, 751–757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kapogiannis D et al. (2019) Association of Extracellular Vesicle Biomarkers With Alzheimer Disease in the Baltimore Longitudinal Study of Aging. JAMA Neurology 76, 1340–1351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Porro C et al. (2019) The multiple roles of exosomes in Parkinson’s disease: an overview. Immunopharmacology and Immunotoxicology 41, 469–476 [DOI] [PubMed] [Google Scholar]
- 49.Surgucheva I et al. (2012) γ-Synuclein: seeding of α-synuclein aggregation and transmission between cells. Biochemistry 51 (23), 4743–4754 [DOI] [PubMed] [Google Scholar]
- 50.Sackmann V et al. (2019) Inhibition of nSMase2 reduces the transfer of oligomeric α-synuclein irrespective of hypoxia. Frontiers in Molecular Neuroscience 12, 200–200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tsutsumi R et al. (2019) Involvement of exosomes in dopaminergic neurodegeneration by microglial activation in midbrain slice cultures. Biochemical and Biophysical Research Communications 511, 427–433 [DOI] [PubMed] [Google Scholar]
- 52.Niu M et al. (2020) A longitudinal study on α-synuclein in plasma neuronal exosomes as a biomarker for Parkinson’s disease development and progression. Eur J Neurol 27, 967–974 [DOI] [PubMed] [Google Scholar]
- 53.Thompson AG et al. (2016) Extracellular vesicles in neurodegenerative disease – pathogenesis to biomarkers. Nature Reviews Neurology 12, 346–357 [DOI] [PubMed] [Google Scholar]
- 54.Pehar M et al. (2007) Mitochondrial superoxide production and nuclear factor erythroid 2-related factor 2 activation in p75 neurotrophin receptor-induced motor neuron apoptosis. Journal of Neuroscience 27 (29), 7777–7785 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Iguchi Y et al. (2016) Exosome secretion is a key pathway for clearance of pathological TDP-43. Brain 139 (Pt 12), 3187–3201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Vella LJ et al. (2007) Packaging of prions into exosomes is associated with a novel pathway of PrP processing. Journal of Pathology 211, 582–590 [DOI] [PubMed] [Google Scholar]
- 57.Guo BB et al. (2015) The neutral sphingomyelinase pathway regulates packaging of the prion protein into exosomes. Journal of Biological Chemistry 290, 3455–3467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Vilette D et al. (2015) Efficient inhibition of infectious prions multiplication and release by targeting the exosomal pathway. Cellular and Molecular Life Sciences 72 (22), 4409–4427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yoo SW et al. (2020) Inhibition of neutral sphingomyelinase 2 promotes remyelination. Sci Adv 6 (40) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Matsuzaka Y et al. (2020) The nSMase2/Smpd3 gene modulates the severity of muscular dystrophy and the emotional stress response in mdx mice. BMC Medicine 18, 343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yoo S-W et al. (2020) Inhibition of neutral sphingomyelinase 2 promotes remyelination. Science Advances 6 (40), eaba5210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Guo BB et al. (2016) Stimulating the release of exosomes increases the intercellular transfer of prions. Journal of Biological Chemistry 291, 5128–5137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Clarke CJ (2018) Neutral sphingomyelinases in cancer: friend or foe? Advances in Cancer Research 140, 97–119 [DOI] [PubMed] [Google Scholar]
- 64.Guarino BD et al. (2019) Extracellular vesicles from pathological microenvironment induce endothelial cell transformation and abnormal angiogenesis via modulation of TRPV4 channels. Frontiers in Cell and Developmental Biology 7, 344–344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ohshima K et al. (2014) Exosome-mediated extracellular release of polyadenylate-binding protein 1 in human metastatic duodenal cancer cells. Proteomics 14 (20), 2297–2306 [DOI] [PubMed] [Google Scholar]
- 66.Takikawa T et al. (2017) Exosomes derived from pancreatic stellate cells: microRNA signature and effects on pancreatic cancer cells. Pancreas 46, 19–27 [DOI] [PubMed] [Google Scholar]
- 67.Matsumoto A et al. (2017) Accelerated growth of B16BL6 tumor in mice through efficient uptake of their own exosomes by B16BL6 cells. Cancer Science 108, 1803–1810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang B et al. (2019) Colorectal cancer cell-derived exosomes promote proliferation and decrease apoptosis by activating the ERK pathway. International Journal of Clinical and Experimental Pathology 12, 2485–2495 [PMC free article] [PubMed] [Google Scholar]
- 69.Huang Z et al. (2018) Exosomes derived from hypoxic colorectal cancer cells transfer Wnt4 to normoxic cells to elicit a prometastatic phenotype. International Journal of Biological Sciences 14 (14), 2094–2102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sang L et al. (2018) Mifepristone inhibits the migration of cervical cancer cells by inhibiting exocrine secretion. Pharmacology 101 (5-6), 322–329 [DOI] [PubMed] [Google Scholar]
- 71.You J et al. (2019) Snail1-dependent cancer-associated fibroblasts induce epithelial-mesenchymal transition in lung cancer cells via exosomes. QJM: An International Journal of Medicine 112, 581–590 [DOI] [PubMed] [Google Scholar]
- 72.Zheng P. et al. (2018) Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death & Disease 9, 434–434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Xu Z-H et al. (2019) Brain microvascular endothelial cell exosome-mediated S100A16 up-regulation confers small-cell lung cancer cell survival in brain. FASEB Journal 33, 1742–1757 [DOI] [PubMed] [Google Scholar]
- 74.Wu B. et al. (2019) Exosomes isolated from CAPS1-overexpressing colorectal cancer cells promote cell migration. Oncology Reports 42, 2528–2536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Richards KE et al. (2017) Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 36 (13), 1770–1778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kavanagh EL et al. (2017) Protein and chemotherapy profiling of extracellular vesicles harvested from therapeutic induced senescent triple negative breast cancer cells. Oncogenesis 6, e388–e388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liu T et al. (2017) Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochimica et Biophysica Sinica 49, 808–816 [DOI] [PubMed] [Google Scholar]
- 78.Faict S et al. (2018) Exosomes play a role in multiple myeloma bone disease and tumor development by targeting osteoclasts and osteoblasts. Blood Cancer Journal 8, 105–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gu YY et al. (2019) Suppressing the secretion of exosomal miR-19b by gw4869 could regulate oxaliplatin sensitivity in colorectal cancer. Neoplasma 66, 39–45 [DOI] [PubMed] [Google Scholar]
- 80.Liu X et al. (2019) Exosomal transfer of miR-501 confers doxorubicin resistance and tumorigenesis via targeting of BLID in gastric cancer. Cancer Letters 459, 122–134 [DOI] [PubMed] [Google Scholar]
- 81.Peak TC et al. (2020) Syntaxin 6-mediated exosome secretion regulates enzalutamide resistance in prostate cancer. Molecular Carcinogenesis 59, 62–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kong JN et al. (2015) Guggulsterone and bexarotene induce secretion of exosome-associated breast cancer resistance protein and reduce doxorubicin resistance in MDA-MB-231 cells. International Journal of Cancer 137, 1610–1620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Chen G et al. (2018) Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560 (7718), 382–386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yang Y et al. (2018) Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Research 28, 862–864 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Poggio M et al. (2019) Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 177, 414–427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kulshreshtha A et al. (2013) Proinflammatory role of epithelial cell-derived exosomes in allergic airway inflammation. Journal of Allergy and Clinical Immunology 131, 1194–1203.el 114 [DOI] [PubMed] [Google Scholar]
- 87.Gon Y et al. (2017) Selective release of miRNAs via extracellular vesicles is associated with house-dust mite allergen-induced airway inflammation. Clinical & Experimental Allergy 47, 1586–1598 [DOI] [PubMed] [Google Scholar]
- 88.Okuro RT et al. (2018) The role of sphingolipid metabolism disruption on lipopolysaccharide-induced lung injury’ in mice. Pulmonary Pharmacology & Therapeutics 50, 100–110 [DOI] [PubMed] [Google Scholar]
- 89.Aslan M. et al. (2014) Inhibition of neutral sphingomyelinase decreases arachidonic acid mediated inflammation in liver ischemia-reperfusion injury’. International Journal of Clinical and Experimental Pathology 7, 7814–7823 [PMC free article] [PubMed] [Google Scholar]
- 90.Gao G et al. (2020) Glutaminase 1 Regulates neuroinflammation after cerebral ischemia through enhancing microglial activation and pro-inflammatory’ exosome release. Frontiers in Immunology 11, 161–161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gu L et al. (2013) Early activation of nSMase2/ceramide pathway in astrocytes is involved in ischemia-associated neuronal damage via inflammation in rat hippocampi. Journal of Neuroinflammation 10, 879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Coblentz PD et al. (2019) Small-hairpin RNA and pharmacological targeting of neutral sphingomyelinase prevent diaphragm weakness in rats with heart failure and reduced ejection fraction. American journal of physiology. Lung Cellular and Molecular Physiology 316, L679–L690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Essandoh K et al. (2015) Blockade of exosome generation with GW4869 dampens the sepsis-induced inflammation and cardiac dysfunction. Biochimica et Biophysica Acta 1852, 2362–2371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ohanian J et al. (2012) Endothelin-1 stimulates small artery VCAM-1 expression through p38MAPK-dependent neutral sphingomyelinase. Journal of Vascular Research 49, 353–362 [DOI] [PubMed] [Google Scholar]
- 95.Gao W et al. (2016) Exosomes derived from mature dendritic cells increase endothelial inflammation and atherosclerosis via membrane TNF-α mediated NF-κB pathway. Journal of Cellular and Molecular Medicine 20, 2318–2327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Lallemand T et al. (2018) nSMase2 (type 2-neutral sphingomyelinase) deficiency or inhibition by GW4869 reduces inflammation and atherosclerosis in Apoe(−/−) mice. Arteriosclerosis, Thrombosis, and Vascular Biology 38, 1479–1492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lu Z et al. (2019) Amitriptyline inhibits nonalcoholic steatohepatitis and atherosclerosis induced by high-fat diet and LPS through modulation of sphingolipid metabolism. American Journal of Physiology Endocrinology and Metabolism 318, E131–E144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Supinski GS et al. (2015) Neutral sphingomyelinase 2 is required for cytokine-induced skeletal muscle calpain activation. American Journal of Physiology. Lung Cellular and Molecular Physiology 309, L614–L624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Caobi A et al. (2020) Extracellular vesicles in the pathogenesis of viral infections in humans. Viruses 12, 1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Chiozzini C et al. (2017) Trans-dissemination of exosomes from HIV-1-infected cells fosters both HIV-1 trans-infection in resting CD4+ T lymphocytes and reactivation of the HIV-1 reservoir. Archives of Virology 162, 2565–2577 [DOI] [PubMed] [Google Scholar]
- 101.Huang Y et al. (2018) Zika virus propagation and release in human fetal astrocytes can be suppressed by neutral sphingomyelinase-2 inhibitor GW4869. Cell Discovery 4, 19–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zhou W et al. (2019) Exosomes mediate Zika virus transmission through SMPD3 neutral Sphingomyelinase in cortical neurons. Emerging Microbes & Infections 8, 307–326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Grünvogel O et al. (2018) Secretion of hepatitis C virus replication intermediates reduces activation of toll-like receptor 3 in hepatocytes. Gastroenterology 154, 2237–225 l.e2216 [DOI] [PubMed] [Google Scholar]
- 104.Nagashima S et al. (2014) Hepatitis E virus egress depends on the exosomal pathway, with secretory exosomes derived from multivesicular bodies. Journal of General Virology 95, 2166–2175 [DOI] [PubMed] [Google Scholar]
- 105.Zhou W et al. (2018) Exosomes serve as novel modes of tick-borne flavivirus transmission from arthropod to human cells and facilitates dissemination of viral RNA and proteins to the vertebrate neuronal cells. PLoS Pathogens 14, e1006764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Vora A et al. (2018) Arthropod EVs mediate dengue virus transmission through interaction with a tetraspanin domain containing glycoprotein Tsp29Fb. Proceedings of the National Academy of Sciences of the United States of America 115 (28), E6604–E6613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wang J et al. (2019) Exosomes released from rabies virus-infected cells may be involved in the infection process. Virol Sin 34, 59–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Streck NT et al. (2020) Human cytomegalovirus utilizes extracellular vesicles to enhance virus spread. J Virol. Published online July 30, 2020. 10.1128/JVI.00609-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Xu X et al. (2019) Detection of viral components in exosomes derived from NDV-infected DF-1 cells and their promoting ability in virus replication. Microbial Pathogenesis 128, 414–422 [DOI] [PubMed] [Google Scholar]
- 110.Takagishi T et al. (2016) Oligomer formation of Clostridium perfringens epsilon-toxin is induced by activation of neutral sphingomyelinase. Biochimica et Biophysica Acta Biomembranes 1858, 2681–2688 [DOI] [PubMed] [Google Scholar]
- 111.Lee K-S et al. (2020) Exosomes released from Shiga toxin 2a-treated human macrophages modulate inflammatory responses and induce cell death in toxin receptor expressing human cells. Cellular Microbiology 22, e13249. [DOI] [PubMed] [Google Scholar]
- 112.Wooff Y et al. (2020) Small-medium extracellular vesicles and their miRNA cargo in retinal health and degeneration: mediators of homeostasis, and vehicles for targeted gene therapy. Front Cell Neurosci 14, 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yang R et al. (2019) Exosomes derived from M2b macrophages attenuate DSS-induced colitis. Front Immunol 10, 2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Elia CA et al. (2019) Extracellular vesicles from mesenchymal stem cells exert pleiotropic effects on amyloid-beta, inflammation, and regeneration: a spark of hope for Alzheimer’s disease from tiny structures? Bioessays 41, e1800199. [DOI] [PubMed] [Google Scholar]
- 115.Monguió-Tortajada M et al. (2017) Nanosized UCMSC-derived extracellular vesicles but not conditioned medium exclusively inhibit the inflammatory response of stimulated T cells: implications for nanomedicine. Theranostics 7, 270–284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Royo F et al. (2020) Methods for separation and characterization of extracellular vesicles: results of a worldwide survey performed by the ISEV Rigor and Standardization Subcommittee. Cells 9, 1955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.You Y et al. (2020) Activated human astrocyte-derived extracellular vesicles modulate neuronal uptake, differentiation and firing. J Extracell Vesicles 9, 1706801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Tabatadze N et al. (2010) Inhibition of neutral sphingomyelinase-2 perturbs brain sphingolipid balance and spatial memory in mice. J Neurosci Res 88 (13), 2940–2951 [DOI] [PMC free article] [PubMed] [Google Scholar]

